EP2055316A1 - Approches basées sur un vecteur lentivirus pour la génération d'une réponse immunitaire au VIH chez les humains - Google Patents

Approches basées sur un vecteur lentivirus pour la génération d'une réponse immunitaire au VIH chez les humains Download PDF

Info

Publication number
EP2055316A1
EP2055316A1 EP08020274A EP08020274A EP2055316A1 EP 2055316 A1 EP2055316 A1 EP 2055316A1 EP 08020274 A EP08020274 A EP 08020274A EP 08020274 A EP08020274 A EP 08020274A EP 2055316 A1 EP2055316 A1 EP 2055316A1
Authority
EP
European Patent Office
Prior art keywords
vector
hiv
vectors
virus
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP08020274A
Other languages
German (de)
English (en)
Inventor
Xiaobin Lu
Boro Dropulic
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
VirxSys Corp
Original Assignee
VirxSys Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by VirxSys Corp filed Critical VirxSys Corp
Publication of EP2055316A1 publication Critical patent/EP2055316A1/fr
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/21Retroviridae, e.g. equine infectious anemia virus
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5256Virus expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5258Virus-like particles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/20011Rhabdoviridae
    • C12N2760/20211Vesiculovirus, e.g. vesicular stomatitis Indiana virus
    • C12N2760/20234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the present invention relates to multiple novel approaches for the generation of an immune response in humans using lentivirus-based vector technology.
  • the invention provides for the ability to mimic the efficacy of a live attenuated (LA) vaccine, without exposing the patient to the risk of disease, as previously demonstrated with LA HIV vaccines for example.
  • LA live attenuated
  • the invention prevents viral escape that has resulted in the failure of previous vaccines due the extreme changeable nature of viruses such as HIV.
  • the control against escape is provided via use of lentiviral vector-based technology to present multiple antigens, preferably as found in, or approximating, wildtype occurrences of the antigens in vivo.
  • the presentation of multiple antigens provides for the generation of a diversified immune response.
  • the lentiviral based vectors include conditionally replicating vectors as well as those that produce non-infectious vector-like particles (VLPs). While the invention is exemplified with respect to HIV, the strategies may be readily applied to the generation of immune responses against other viruses or microorganisms, including bacteria.
  • HAART highly active antiretroviral therapy
  • NRTI nucleoside reverse transcriptase inhibitor
  • NRTI non-nucleoside reverse transcriptase inhibitor
  • PI protease inhibitor
  • vaccines provide protection from virus infection by eliciting a strong antiviral neutralizing antibody response.
  • Neutralizing antibodies recognize proteins on the virus surface and prevent binding to and infection of healthy cells.
  • This approach is not effective against HIV due to the broad range of HIV subtypes and rapid mutation rate that allows HIV to escape immune responses that are not sufficiently diverse.
  • the most successful of vaccines designed to elicit neutralizing antibody are bivalent vaccines that consist of recombinant envelope proteins derived from two different strains of HIV. VaxGen has led the field with these bivalent vaccines, but although some protective immunity is elicited, the immune response to these vaccines remains poor.
  • cytotoxic T-lymphocytes CTLs
  • CD8+ T lymphocytes CD8+ T lymphocytes
  • LA vaccines do not cause disease in humans, but they are able to replicate and elicit a broad, well-rounded immune response consisting of both cellular immunity and a neutralizing antibody response to HIV proteins produced during infection. It is especially important that a diverse immune response be mounted against HIV since the high mutation rate of the virus during infection changes how the virus looks to the immune system thus contributing to immune evasion.
  • a LA vaccine presents these diverse variables to the immune system thereby avoiding this problem.
  • an attenuated HIV vaccine is represented by a group of people who were inadvertently infected with an attenuated (delta-Net) strain of HIV through a blood infusion from an infected individual ( Deacon N.J. et al. Genomic structure of an attenuated quasi species of HIV-1 from a blood transfusion donor and recipients. Science. 270, 988-991 (1995 ); Learmont J.C. et al. Immunologic and virologic status after 14 to 18 years of infection with an attenuated strain of HIV-1. A report from the Sydney Blood Bank Cohort. N. Engl. J. Med. 340, 1715-1722. (1999 )).
  • LTNPs long term non-progressors
  • LTNPs The presence of LTNPs among HIV patients indicates that host and viral factors influence the pathogenesis of HIV/AIDS. Specifically, persons deficient in the chemokine receptor CCR5 fail to become infected with HIV. The patients discussed earlier who were infected with naturally acquired attenuated HIV viral genomes (nef-deleted) also exhibited delayed progression to AIDS.
  • New diseases are continually emerging at least each decade, such as HIV in the 1980s, EBOLA in the 1990s, and SARS in the 2000s.
  • a new disease emerges, significant amounts of research resources are devoted to developing a protective vaccine for the disease, as each disease requires different types of vaccines to effect protection. This is the case because some diseases are controlled by humoral responses, some by cellular responses, and some require both.
  • the risk of a live attenuated vaccine must be assessed, as well as the efficacy of a killed virus or recombinant protein vaccine.
  • a vector-based approach to vaccination that elicits diversified cellular and humoral immunity, would allow a single vaccine approach for all diseases that may be re-engineered according to the genetic structure of each emerging threat.
  • the present invention provides multiple novel approaches for the generation of an immune response, preferably against viruses such as HIV, in humans using lentivirus-based vector technology. Without exposing a treated subject to the risk of disease as previously observed with live attenuated (LA) vaccines, the invention provides compositions and methods that are able to mimic or reproduce the efficacy of a LA vaccine. Moreover, the invention addresses the phenomenon of high mutation rates that result in viral escape from the effects of previous vaccines.
  • LA live attenuated
  • the invention provides for lentiviral vector-based technology that utilizes conditionally replicating virus for antigen presentation in vivo .
  • This vector-based technology allows for the expression of multiple antigens to allow for the generation of a diversified immune response, although the response does not necessarily have to be against every antigen or epitope presented by the invention. All that is needed is the generation of a response to one or more antigens expressed by the use of the invention.
  • the response is cellular and humoral in nature, although the occurrence of either response may occur in the practice of the invention. Even more preferred are response(s) that are protective against subsequent challenge with the antigen(s) or pathogens that present the antigen(s).
  • a vector based approach offers several advantages. These include concurrent presentation of multiple antigens; no (or reduced) bifurcation of the immune response between a heterologous virus carrier and the immunogens; mimicking of wild-type virus replication in the context of conditional vector replication; and extended period of antigen production.
  • the invention thus provides for the use of a system of multiple (two or more) complementary, but individually replication-defective vectors, or conditionally replicating viral vectors such as conditionally replicating HIV vectors (crHIVs). This design provides a vaccine vector system that is safer than live attenuated (LA) virus and yet more potent than single replication defective vectors.
  • two complementary, replication-defective HIV-1 vectors can provide limited replication and packaging of both vectors cells based on observations made by the present inventors using more than one vector, one of which expresses a VSV-G envelope protein to complement replication of another vector.
  • the replication of an individual crHIV is always suboptimal since the system requires co-infection of the very same cells with the necessary complementary vectors in order to produce viable virus particles.
  • multiple HIV antigens are produced to elicit robust humoral and cellular immune responses similar to that seen with vaccination using a LA virus vaccine. Each vector alone cannot replicate itself and so cannot spread to new cells.
  • the ability of the vectors to replicate and mutate in a conditional manner thus provides an "active" vaccine that constantly provides new antigenic displays to elicit a robust and broad immune response better able to protect against pathogens, such as HIV, than any of the current alternative vaccines being tested. It should be noted, however, that this active production of variant antigens does not increase the potential for virulence because the underlying vectors are inherently made replication-defective such that more than random mutation of individual coding regions is necessary for restoration of replication competence.
  • any essential viral protein may be provided in trans , and will be utilized by vectors needing it for replication as part of the invention's intended (but non-limiting) therapeutic mechanism.
  • RCVs replication competent vectors
  • molecular designs genetic antiviral agents
  • examples of such agents in each vector include, but are not limited to, targeted antisense sequences, ribozymes, and post-translational gene silencing (PTGS) directed to the other vector(s).
  • PTGS examples include small interfering RNAs (siRNAs) and RNA inhibition (RNAi) as described below.
  • siRNAs small interfering RNAs
  • RNAi RNA inhibition
  • agents may be differentially targeted in the cells.
  • one vector may express an agent that expresses in the nucleus while the agent expressed by the second vector traffics to the cytoplasm.
  • Another non-limiting approach to prevent recombination between the two vectors without curtailing their independent propagation is to place the expression cassette in one of the two complementary vectors in a reverse orientation. Since the likelihood of flipping back to the original orientation through recombination is extremely low, or perhaps impossible, then the possibility of recombination is avoided and targeted agents may not be needed. Viral factors needed in cis for replication, however, will be present on all conditionally replicating vectors that are to be susceptible to packaging and proliferation.
  • Conditionally replicating vectors may be pseudotyped with any suitable envelope protein, including, but not limited to, the VSV-G envelope protein, a native HIV or HTLV envelope, or any molecule that targets CD4+ T lymphocytes and/or macrophages or dendritic cells.
  • the pseudotyped particles may contain at least one copy of each of the vectors necessary to complement replication of them all.
  • the particles may not contain at least one copy of each of the vectors necessary to complement replication of them all, but instead contain less than all of the vectors.
  • a combination of particles providing all of the necessary vectors may still be introduced into cells via the use of particles that can multiply infected the intended cells to provide the necessary combination of particles.
  • each can be separately packaged into particles which are then contacted with target cells for infection under conditions where the cells would be multiply infected with the particles such that some, many, or all of the cells would be infected with at least one copy of each vector.
  • the conditionally replicating vectors may be used to transduce highly concentrated target cells ex vivo using autologous cell transplantation.
  • complementing vectors may be used in vivo , such as by injection made intramuscularly, subdermally, systemically, or in an area targeted for direct drainage into the lymphatic system.
  • Boosters may consist of simple DNA vaccinations given intramuscularly, or subdermally using the DNA of one or more of the conditionally replicating vectors.
  • Alternative boosters by use of other genetic (vector) or proteinaceous vehicles (e.g. vaccines) may also be used to provide complementary factors in trans .
  • delivery of Tat protein, or other vector capable of expressing Tat protein may be used to activate replication of the second vector.
  • the above complementary vector system may also be modified to produce non-infectious virus-like particles (VLPs).
  • the VLPs are lentivirus-like particles, but they may be a hybrid of viral and cellular components that make up the particles, such as, but not limited to the case of particles composed of HIV-1 proteins except for the use of an HIV-2 or other modified or heterologous env protein.
  • the simplest modification is to omit viral factors necessary in cis from one or more of the vectors.
  • a cell containing all the vectors of the system would produce viral particles using all of the necessary (and optionally non-essential) replication and packaging viral proteins, at least one of the vectors would not be capable of being packaged into the particles. This permits control over the propagation, from one cell to another, of the ability to produce VLPs.
  • the coding sequence of one or more viral factors necessary in trans may be mutated or omitted such that only one round of replication and packaging may occur.
  • a non-limiting example in the case of HIV is a mutation in, or deletion (in whole or in part) of, the pol gene that prevents expression of reverse transcriptase and/or protease activity.
  • a vector containing such a mutation or deletion may be packaged in vitro with a helper/packaging vector which provides reverse transcriptase and/or protease activity in trans , which is also incorporated into the resultant particle to permit one round of replication and packaging after introduction into a susceptible cell with the necessary complementary vectors.
  • the invention provides for the production of non-infectious vector-like particles (VLPs) without the use of two or more complementary conditionally replicating vectors.
  • VLPs non-infectious vector-like particles
  • lentiviral vector-based production of antigens simultaneously stimulate cellular and humoral immunity for maximum response against a virus or microbial agent, such as, but not limited to, HIV.
  • a virus or microbial agent such as, but not limited to, HIV.
  • HIV HIV-based vector encoding replication defective-virus like particles (RD-VLPs) is designed to generate both humoral and cell mediated immune responses.
  • This (vaccine) vector may be viewed as "Toti-VacHIV" for its total (or nearly total) presentation of the HIV antigens and ability to stimulate both antibody and cell mediated immune responses. Therefore, the protection provided by this vector is expected to be more comprehensive than traditional single modality (single antigen) vaccines or vaccines that present limited numbers of antigens.
  • the invention contemplates the use of "nearly total” vectors that present less than all possible antigens, and antigenic epitopes, of HIV or any other targeted virus or microorganism.
  • Toti-VacHIV is designed to express epitopes from each viral protein as they would be processed by cells in vivo.
  • Toti-VacHIV is a lentiviral based vector comprising both 5' and 3' LTR elements and a heterologous (to HIV or other lentiviral vector) constitutively active promoter that directs expression of HIV encoded gene products after introduction into a host or target cell. This may be following conversion of the vector into a DNA form via reverse transcriptase activity.
  • the choice of a constitutively active promoter may be any preferred by the skilled person, including, but not limited to, that derived from simian cytomegalovirus (S-CMV-P).
  • a heterologous inducible promoter may be used in place of a constitutively active promoter.
  • the epitopes in the case of HIV and other lentiviruses and some retroviruses, may include, but are not limited to, those from the gag-pol, vif, vpr, and env regions, derived from the unspliced or partially spliced messenger RNA, as well as the epitopes from the tat, rev, and nef regions, which are derived from the multiply spliced mRNA. De novo synthesis of these antigens in cells containing the vector, such as vaccinated antigen presenting cells, are directed to the MHC class I pathway for generation of cell mediated immune responses.
  • VLPs beneficial feature of this vector, as well as the complementary vectors discussed above for the production of VLPs, is the ability to make completely defective VLPs by deleting of some of the functionally vital regions of gag-pol and env structural genes such that the vector(s) retain the ability to result in assembly of the VLPs in cells.
  • the VLPs that are released from cells containing the vector(s) will be able to induce antibody responses.
  • An advantage to the VLPs is that some of the antibodies generated will be directed to the viral conformational epitopes as found on the surface of viral particles and thus effect virus neutralization (see Figure 2 ).
  • sizeable mutations may be introduced in critical regions of viral genes and elements (e.g. cis -acting elements). Multiple deletions in the viral sequence will dramatically reduce the possibility of reversion of the vector to a replication competent virus.
  • the cis-acting packaging signal ( ⁇ ), primer binding site (PBS), the central polypurine tract (cPPT) and/or the polypurine tract (PPT) are all removed from the vector to prevent it from being packaged and/or reverse transcribed.
  • functional regions encoding reverse transcriptase (RT) and/or integrase (IN) in the pol region, and envelope (env) structural genes may be deleted to wholly ensure defective replication of the vector.
  • the gag function for VLP assembly is preserved to ensure the production of the defective particles for generation of an antibody response as described above (see Figure 3 ).
  • portions of the RT, IN and/or env encoding sequences may be retained if immune responses directed at the epitopes encoded by those portions are desired.
  • such portions do not encode the normal functions of RT, IN or env beyond those necessary to produce VLPs.
  • Non-limiting methods for delivery of the vaccine vector to a subject or patient include, but are not limited to, the intramuscular, subdermal, or systemic route using naked DNA in the presence of an adjuvant, followed with booster injections using Toti-VacHIV DNA and VLPs generated in culture (see Figure 4 ).
  • the vaccine may be given via ex vivo transduction of target cells, most preferably lymphocytes, and/or macrophages or dendritic cells (or other antigen presenting cells) using vectors packaged in a lentiviral packaging system that pseudotypes the vector with an appropriate envelope protein, such as, but not limited to, the G protein from vesicular stomatitis virus (VSV).
  • VSV vesicular stomatitis virus
  • the use of multi-antigen expressing vectors is provided.
  • Person to person variation in antigen recognition results primarily from the polymorphism present in the peptide binding site of the major histocompatibility complex (MHC) class I and II molecules, which function to present foreign antigens to T and B cells, respectively, to result in generation and evolution of the immune response to the presented antigens.
  • MHC major histocompatibility complex
  • the specific peptide presented will differ depending upon the conformation of the peptide binding site in the MHC molecule. Therefore, a vaccine applicable across a broad human population will preferably use several peptide antigens to ensure effective stimulation among most or all subjects treated.
  • LTNPs long term non-progressors
  • the epitopes that are recognized by a more developed host immune system can be divided into 3 major classes according to the responding cells: cytotoxic T lymphocytes (CTL), helper CD4+ T lymphocytes (CD4), and B cells.
  • CTL cytotoxic T lymphocytes
  • CD4 helper CD4+ T lymphocytes
  • B cells B cells.
  • the B cell epitopes may be further categorized into conformational and linear epitopes depending upon whether the epitope is recognized as a three dimensional native structure, or as a denatured linear entity.
  • a vast number of immune dominant epitopes have been experimentally defined in conjunction with their associated MHC genes, and may be easily expressed as part of a multi-antigen presentation by a vector of the invention.
  • the invention provides for any of the vectors disclosed herein to contain a comprehensive spectrum of epitopes representing different HLA types.
  • the invention can also be used to functionally determine epitopes for various disease-causing infectious agents, including HIV-1 and HIV-2, in the context of MHC restriction.
  • the advantage of this approach over the existing synthetic peptide is that native and naturally, rather than artificially, processed epitopes are selected.
  • a disadvantage of previous peptide vaccines is the difficulty to produce synthetic molecules mimicking conformational epitopes (immunological determinants in the native protein which are formed by amino acid residues brought together as part of protein folding)
  • the vectors of the present invention are designed to produce proteins and antigens in a more native context. Multiple members of the identified antigens and epitopes may then be combined for expression in the vectors of the invention.
  • Such a multi-antigen vector contains one or more sequences encoding many conserved dominant epitopes for stimulation of (and recognition by) CTL, CD4+ and/or B cells linked together as one or more polypeptides. Preferably inserted between the epitopes are conserved peptide antigen processing sequences.
  • the epitopes are designed to cover each viral protein for development of a multivalent vaccine designed to reduce the likelihood of virus mutants capable of escaping the ensuing immune response. Stated differently, the epitopes are preferably those conserved in many or all strains of a virus or other pathogen.
  • the identification or determination of various epitopes as potentiating CTL, CD4+ and/or B cell responses, particularly strong and protective responses may be by any method known in the art. Such epitopes, or appropriately representative members thereof, are preferably used in the practice of the invention to stimulate a strong and protective cellular and humoral immune response in the subject.
  • the above strategies maybe used independently, or in combination with each other and/or with other vaccine strategies known to the skilled person, for administration to a subject in need thereof.
  • Such subjects include individuals who are already infected with a virus or microbial agent, such as HIV, as well as individuals at risk for such infections.
  • Administration may be by any means known in the art, including, but not limited to contacting one or more cells of said subject with the compositions of the invention.
  • the disclosed strategies may be easily reengineered to generate immune responses, and protective states of vaccination, against many other viral or bacterial infections, or cancer.
  • Preferred subjects for the practice of the invention are humans, although the invention may be adapted for use in other organisms, particularly mammals and primates.
  • the invention provides compositions such as the above nucleic acid constructs and vectors, viral particle encapsulated forms thereof, formulations for their use, and methods of their use to generate immune responses, and protective states of vaccination, against many other viral or bacterial infections, or cancer. Additionally, the invention may be embodied in the form of kits comprising components of the invention for the practice of methods disclosed herein.
  • Figure 1 shows a schematic of one embodiment as representation of the strategy of the present invention using a system of complementary, replication deficient vectors.
  • Step 1 intravenous delivery of vectors to T cells; first round of infection in cells.
  • Step 2 production of 1 st round of vector; co-packaged vector suppressed by antisense safety feature.
  • Step 3 second round of infection in T cells; co-infection yields 2 nd round of vector production.
  • Step 4 propagation of vector infection in cells until immune-mediated clearance.
  • FIG. 2 shows a schematic of one embodiment as representation of the strategy of the present invention using a viral like particles (VLPs).
  • VLPs viral like particles
  • Figure 3 shows one embodiment of the invention comprising the genome of a replication defective HIV vector to produce VLPs.
  • All epitope peptides are defective by sizable deletions to prevent revertants. Epitopes are expressed from their original configuration in the viral genome.
  • E GP is defective in RT and IN but able to assemble budding-able virus-like particles.
  • E vif-Vpr contains conservative epitopes.
  • E tat delete the essential R-domain for TAR which is not immunogenic.
  • E env delete the protease cleavage site between gp120 and gp41.
  • E Nef contains deletions in RR, Myr, NBP-1, ⁇ -COP binding sites. Rev is the only functional viral protein in this system.
  • Defective cis-acting elements -TAR, - ⁇ , -PBS, -cPPT, -PPT.
  • Figure 3 depicts a complete replication-defective viral DNA vector with the following features: presentation of many epitopes with a single vector and antigen presentation via MHCI for CTL (endogenous epitope expression) and MHCII for antibody (virus- like particles released from vaccinated cells, epitopes can be in their native conformation).
  • Figure 4 shows one embodiment of the invention comprising the use of a replication defective HIV vector and VLPs to product an immune response.
  • Figure 5 shows the propagation of a system of complementary, replication deficient HIV-based Vectors in Cell Culture.
  • a lentiviral vector minimally contains LTRs from a lentivirus and optionally packaging sequences in the 5' leader and gag encoding sequences of a lentivirus.
  • the vector may also optionally contain the RRE element to facilitate nuclear export of vector RNA in a Rev dependent manner.
  • Each of the aspects of the invention described above and herein is designed to maximize the likelihood of that the immune response generated by their use will contain the diversity of an incoming pathogen, such as the HIV virus, by generating (1) a multivalent response to viral antigens that mimics the diversity among naturally mutating HIV; (2) a broad breadth of immune stimulation; and (3) a balanced humoral and cellular immune response. It is worthy to note that in a TotiVacHIV system, the native conformations of viral proteins are preserved in the context of mature virus particles, which may provide an advantage for production of neutralizing antibodies.
  • a patient may first be immunized with a multi-antigen vector followed by a system of complementary, conditionally replicating vectors to facilitate diversification of the response.
  • these vaccine approaches may be combined with previously tested vaccines known to the skilled person, such as a LA vaccine, a killed vaccine, or a single protein (or other recombinant) vaccine.
  • a patient may be immunized first with Toti-VacHIV to prime immunity so that he/she may be subsequently vaccinated with live attenuated HIV for development of a powerful and diverse immune response without disease.
  • Figure 1 illustrates an embodiment of the invention in which two complementary, conditionally replicating vectors, VRX-V2A and VRX-V2B, are used to generate an immune response by stimulating T cells or dendritic cells upon introduction.
  • each vector alone cannot propagate (one encodes the structural proteins needed in trans while the other encodes the non-structural proteins needed in trans ), but during co-infection of T lymphocytes they support the replication of one another.
  • Infection of susceptible mammalian cells with the vaccine vectors results in expression of HIV proteins and subsequent stimulation of immunity. During infection by only one vector in a cell, the HIV proteins encoded by the vector are still expressed, but no progeny vector is produced.
  • Methods of introducing vectors into cells are known in the art and may be used in the practice of the invention. As a non-limiting example, the methods include those disclosed in allowed U.S. Patent Application 09/653,088, filed August 31, 2000 , which is hereby incorporated by reference as if fully set forth.
  • RNA RNA sequences to generate polynucleotides for post-transcriptional gene silencing (PTGS).
  • PTGS post-transcriptional gene silencing
  • dsRNA homologous double stranded RNA
  • RNA interference mediated by the directed introduction of dsRNA.
  • Another form is via the use of small interfering RNAs (siRNAs) of less than about 30 nucleotides in double or single stranded form that induce PTGS in cells.
  • siRNAs small interfering RNAs
  • a single stranded siRNA is believed to be part of an RNA-induced silencing complex (RISC) to guide the complex to a homologous mRNA target for cleavage and degradation.
  • RISC RNA-induced silencing complex
  • siRNAs induce a pathway of gene-specific degradation of target mRNA transcripts.
  • siRNAs may be expressed in via the use of a dual expression cassette encoding complementary strands of RNA, or as a hairpin molecule.
  • Steps 3 and 4 of Figure 1 illustrate further rounds of infection and propagation of vaccine vectors, which continues until immune-mediated clearance occurs, commensurate with protective immunity mediated by both cellular and humoral responses.
  • the complementary vectors disclosed herein may be modified to express, and thus generate immune responses, against non-HIV proteins, such as proteins from other viruses or microorganisms.
  • non-HIV proteins such as proteins from other viruses or microorganisms.
  • viruses include other lentiviruses (including HIV-1, HIV-2, EIAV, VMV, CAEV, BIV, FIV, and SIV), retroviruses, and other viruses with envelope glycoproteins, including, but not limited to, togaviruses, rhabdoviruses, paramyxoviruses, herpesviruses, orthomyxoviruses and coronaviruses.
  • a heterologous envelope protein is to be encoded and expressed by a vector, it is preferably one that is capable of pseudotyping the vector.
  • Non-limiting examples of suitable envelope proteins for pseudotyping include the HIV-1, HIV-2, or MMLV envelope protein; the G protein from Vesicular Stomatitis Virus (VSV), Mokola virus, or rabies virus; GaLV; Alphavirus E1/2 glycoprotein; the envelope protein from human T cell leukemia virus (HTLV); RD114, an env protein from feline endogenous virus; or the glycoproteins from other lentiviruses or retroviruses such as gp90 from equine infectious anemia virus (EIAV) or the surface glycoprotein of bovine immunodeficiency virus (BIV).
  • VSV Vesicular Stomatitis Virus
  • HTLV human T cell leukemia virus
  • RD114 an env protein from feline endogenous virus
  • gp90 from equine infectious anemia virus (EIAV) or the surface glycoprotein of bovine immunodeficiency virus (BIV).
  • Sequences encoding an envelope protein from the following viral families may also be used: Piconaviridae, Tongaviridae, Coronaviridae, Rhabdoviridae, paramyxoviridar, Orthomixoviridae, Bunyaviridae, Arenaviridae, Paroviridae, Poxviridae, hepadnaviridae, and herpes viruses.
  • hybrid envelope proteins comprising portions of more than one envelope protein may be encoded and expressed by vectors of the invention.
  • viral envelope proteins are encompassed by the invention.
  • they may be considered envelope protein replacement strategies and are particularly attractive because of the variability of env proteins among viruses, especially HIV.
  • the replacement of the env encoding sequences in a system of HIV or lentiviral vectors with a variant gene from another HIV strain or isolate better tailors the resulting vectors to generate an immune response.
  • lentiviral viruses such as HIV.
  • These alternative vectors may be used to vaccinate against HIV strains which are prevalent in a particular part of the world or in a particular population or against particularly virulent strains.
  • the vectors may also be tailored for therapeutic treatment of a particular infected patient to prevent the onset of AIDS symptoms, for example, by inclusion of sequences encoding the env protein from the particular strain of HIV infecting the patient after its isolation and identification from the patient.
  • env sequence is readily accomplished by cloning the env sequence and replacing the env sequence in the vector with the cloned sequence by routine methods such as the polymerase chain reaction (PCR) and other recombinant DNA techniques.
  • PCR polymerase chain reaction
  • combination of vectors encoding a multiplicity of different Env proteins may be used in the practice of the invention.
  • Variant env encoding sequences can also be engineered by mutagenesis and used in the practice of the invention.
  • the vectors are used with (and express) native envelope proteins that are expected or found in the targeted virus(es). In addition to permitting an immune response to be generated against the envelope proteins of the targeted virus(es), this approach reduces or minimizes the possibility of the targeted virus(es) being repackaged with a heterologous envelope protein that would facilitate viral spread.
  • the vectors of the invention may encode or contain anti-viral agents that prevent the packaging of vectors with copies of the targeted virus(es) in a viral particle.
  • the vectors of the invention will contain elements and agents to reduce or minimize the likelihood of recombination between a vector of the invention and a wildtype virus. Non-limiting examples of such agents and elements are provided in U.S. Patent 6,168,953 , and allowed U.S. Patent Application 09/667,893, filed September 22, 2000 , both of which are hereby incorporated by reference as if fully set forth.
  • the vectors may also encode other viral proteins, including, but not limited to, capsid proteins from other lentiviruses or other retroviruses. Indeed, virtually any protein, structural or non-structural, of a virus or microorganism which may generate a helpful immune response may be expressed by the vectors and methods of the invention as long as they do not prevent replication and/or gene expression from the vector. These proteins may also be subject to the replacement technique described above for env encoding sequences. Moreover, and as would be clear to the skilled person, the vectors of the present invention may be based upon the genomes of other lentiviral vectors as disclosed herein.
  • lentiviral based vectors may also be used to address situations where expression of a heterologous protein interferes with vector replication and/or gene expression in that the interference may be lessened or not present when another lentiviral genome is used.
  • the invention also provides for the expression of proteins that provide an immune response, and/or a protective effect, against the virus that causes SARS (Severe Acute Respiratory Syndrome).
  • vectors based upon HIV may be constructed with mutations of the gp160 cleavage site which block the processing of the gp160 envelope precursor to gp120 and gp41. This may reduce the "shedding" of gp120 antigen observed during propagation of HIV based particles.
  • the gp41 portion of gp160 is a transmembrane peptide which may tether the gp120 antigen more firmly to the membrane in which gp160 is inserted.
  • the increased retention of the gp120 portion should improve the immunogenic properties of the vectors of the invention, and may be adapted to any other antigen where inhibition of "shedding" or increased retention in membranes may be useful in generating an immune response.
  • the env protein may be a chimeric glycoprotein, such as one having elements from both HIV-1 and HIV-2, or having a portion from the V3 loop of the MN viral isolated at various positions within the HIV-1 env gene (as described in USP 5,866,137 ).
  • FIG. 2 shows an embodiment of the invention wherein a replication defective vector that produces HIV virus like particles (VLPs) is used to induce cellular and/or humoral responses against HIV proteins.
  • the vector such as Toti-VacHIV, stably integrates into the genome of the host cell, and produces HIV antigens which are processed internally in the endoplasmic reticulum (ER) and expressed via the MHC class I pathway for stimulation of cellular (CD8 or cytotoxic) immune response. Additionally, proteins are produced by the integrated vector to allow formation of VLPs (via "budding" for example) that are not capable of propagating the vector.
  • the VLPs remain capable, however, of being taken up by cells, such as antigen presenting cells (APCs), and processed by cellular processes therein, including those that contribute to the generation of an immune response.
  • APCs antigen presenting cells
  • FIG 2 A non-limiting example of this is shown in Figure 2 , wherein an APC presents antigens from the VLP via the MHC class II pathway for stimulation of the humoral response.
  • Figure 3 shows the overall design of a possible embodiment of a Toti-VacHIV vector of the invention.
  • the vector is designed to present several HIV epitopes (denoted by "E") after being (stably) introduced into a target cell. Epitopes are separated by splice sites (splice donor or "SD” and splice acceptor or "SA” sites) derived from HIV. The sites shown are “SD1", “SD2", “SA1” and “SA2".
  • the mRNA is driven off of a constitutively expressed promoter, in this case the CMV promoter. Other promoters may be used in the practice of the invention. Non-limiting examples include the Tk promoter, the EF- ⁇ promoter, and the PGK promoter.
  • RNA packaging
  • nuclear import central polypurine tract or cPPT
  • replication primary binding site or PBS
  • other deletions or mutations may be made to inhibit or prevent propagation of the vector (such as those defective in the trans-activated responsive (TAR) region, the gag carboxy-terminal CysHis box (or "zinc knuckle”) region, and polypurine tract within the nef region); and 2) the three exemplary mutations may be used singly, in pairs, or in combination with other deletions or mutations to inhibit or prevent vector propagation.
  • the vector includes sizable deletions in all epitope peptides or antigens; expression of epitopes or antigens based on their native configuration in the HIV viral genome; the gag-pol (GP) epitope is defective in both reverse transcriptase (RT) and integrase (IN) activities but able to assemble VLPs capable of "budding" from a cell; the vif-vpr region retains conserved epitopes; the tat region contains a deletion of the essential, but not known to be immunogenic, R domain for interactions with the trans-activated responsive (TAR) region; the env region contains a deletion of the cleavage site between gp120 and gp41; the nef region contains deletions in RR (double arginine), Myr (myristylation site), NBP-1 (Nef Binding Protein-1), and ⁇ -COP ( ⁇ coatomer protein ) binding sites; and Rev is the only functional viral protein in this embodiment.
  • replication defective vectors of the invention may contain other sequence alterations as well as the ability to express other proteins, antigens and epitopes.
  • proteins from other viruses or pathogenic microbial agents may be expressed by sequences encoding them and placed into the vectors of the invention. In one non-limiting example, this may be by the replacement strategy described above for conditionally replicating vectors.
  • each of such vectors is a complete replication defective viral vector capable of presenting many epitopes concurrently.
  • the epitopes/antigens may be presented via MHC I (via endogenous epitope expression in cells containing the vector) and via MHC II (via VLPs released from cells containing the vector).
  • the epitopes are expected to be in, or closely approximate, their native conformations.
  • a replication deficient vector of the invention may be introduced into susceptible cells, such as, but not limited to, HeLa, HeLa-tat, COS, 293, CHO, BHK, CEMx174, SupT1, Vero cells, 3T3, D17, yeast, bacteria, or primary cells in vivo or ex vivo (particularly of hematopoietic origin) capable of supporting VLP production after transfection with the vector.
  • susceptible cells such as, but not limited to, HeLa, HeLa-tat, COS, 293, CHO, BHK, CEMx174, SupT1, Vero cells, 3T3, D17, yeast, bacteria, or primary cells in vivo or ex vivo (particularly of hematopoietic origin) capable of supporting VLP production after transfection with the vector.
  • the introduction of the vector may be by any known means and may be transient or permanent to result in VLP expression.
  • the VLPs may be isolated from culture supernatant by, as non-limiting examples, pelleting, sucrose gradient purification,
  • the vectors may be introduced into cells of a subject or patient under ex vivo conditions and thereafter returned to the subject or patient.
  • the cells may be confirmed for their ability to produce VLPs before their returned or simply returned to produce VLPs in vivo.
  • the vector may be considered a replication deficient, "proviral" form even though it is replication deficient.
  • Another embodiment of the invention that may be used is where a cell containing the vector and expressing VLPs (i.e. "VLP producer cell”) is introduced into a subject or patient to generate VLPS in vivo . This differs from the ex vivo approach in that the cell need not be necessarily from the subject or patient, in which case there may be an immune response against those cells.
  • the VLPs produced by the above in vitro or ex vivo methods may be introduced into a subject or patient as therapy, thereby obviating the need for the vector to be present in vivo .
  • the use of cells from the subject or patient to be treated is particularly advantageous in that the resultant VLPs may be utilized with a minimum of (or without) issues of rejection.
  • the cells may be maintained ex vivo in culture to produce VLPs for extended periods via techniques known in the art.
  • FIG. 4 shows a representation of a protocol using a replication defective vector mediated method of the invention.
  • the embodiment begins with immunization using Toti-VacHIV DNA.
  • the DNA may be delivered and taken up by cells of a subject by any appropriate means, including cases where the DNA is to be stably integrated into the cell's genome or maintained episomally. Expression of the epitopes by the cells result in presentation thereof in combination in an MHC I context to generate cellular based immune responses.
  • the cells will also be able to express proteins necessary for VLP production followed by their assembly and release as VLPs into the extracellular environment or via direct cell to cell mediated transfer. Uptake of the VLPs by antigen presenting cells results in their presentation in an MHC II context to generate humoral based immune responses.
  • the subject After an appropriate time, the subject would be boosted at least once with a combination of Toti-VacHIV DNA and VLPs (optionally produced by said DNA). This boost augments both the initial cellular and humoral immune responses.
  • the induction or presence of a cellular or humoral response can be assayed at any point based on a chromium release assay for CTL activity and assays for neutralizing antibodies as known to the skilled person.
  • subjects may be boosted separately, or in conjunction with Toti-VacHIV, in combination with another vector disclosed herein or with another vaccine as known in to the skilled person.
  • any of the vectors disclosed herein may be alternatively used to express a multi-antigen construct comprising epitopes identified as of particular interest for the generation of an immune response.
  • the multi-antigen construct preferably includes antigens or epitopes, preferably dominant determinants, which induce both cellular and humoral responses.
  • antigens and epitopes may be identified by use of the vectors of the invention, which can be applied toward presenting various antigens and epitopes singly or in combination in various formats in animal models for the generation of an immune response.
  • Those determinants found to elicit strong cellular and/or humoral responses can be selected and used in the preparation of multi-antigen constructs for expression via the vectors of the invention.
  • a vector or construct of the present invention can be formulated into various compositions to facilitate their use in therapeutic and prophylactic treatment methods.
  • the vectors and constructs can be made into a pharmaceutical composition by combination with appropriate pharmaceutically acceptable carriers or diluents, and can be formulated to be appropriate for either human or veterinary applications. Additionally, formulations for delayed release or release over time are also provided.
  • a composition for use in the method of the present invention can comprise one or more of the aforementioned vectors or constructs, preferably in combination with a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers are well-known to those skilled in the art, as are suitable methods of administration for generation of an immune response. The choice of carrier will be determined, in part, by the particular vector or construct, as well as by the particular method used to administer the composition. The skilled person will also appreciate that various routes of administering a composition are available, and, although more than one route can be used for administration, a particular route can provide a more immediate and more effective reaction than another route. Accordingly, there are a wide variety of suitable formulations of the composition of the present invention.
  • a composition comprised of a vector or construct of the present invention, alone or in combination with other antiviral compounds, can be made into a formulation suitable for direct administration to a subject or administration to a cell of a subject ex vivo .
  • a formulation can include aqueous and nonaqueous, isotonic (or iso-osmotic) sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and nonaqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • the formulations can be presented in unit dose or multidose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water, for injections, immediately prior to use.
  • sterile liquid carrier for example, water
  • Extemporaneously injectable solutions and suspensions can be prepared from sterile powders, granules, and tablets, as described herein.
  • the vector can be stored in any suitable solution, buffer or lyophilizable form, if desired.
  • a preferred storage buffer is Dulbecco's Phosphate Buffered Saline; Dulbecco's Phosphate Buffered Saline mixed with a 1-50% solution of trehalose in water (1:1), preferably a 10% solution of trehalose in water (1:1), such that the final concentration is 5% trehalose; Dulbecco's Phosphate Buffered Saline mixed with a 1-50% solution of glucose in water (1:1), preferably a 10% solution of glucose in water (1:1), such that the final glucose concentration is 5%; 20mM HEPES-buffered saline mixed with 1-50% solution of trehalose in water (1:1), preferably a 10% solution of trehalose in water (1:1), such that the final trehalose concentration is 5%; or; Dulbecco's Phosphate Buffered Saline mixed with
  • a formulation suitable for oral administration can consist of liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or fruit juice; capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as solid or granules; solutions or suspensions in an aqueous liquid; and oil-in-water emulsions or water-in-oil emulsions.
  • diluents such as water, saline, or fruit juice
  • capsules, sachets or tablets each containing a predetermined amount of the active ingredient, as solid or granules
  • solutions or suspensions in an aqueous liquid and oil-in-water emulsions or water-in-oil emulsions.
  • Tablet forms can include one or more of lactose, mannitol, corn starch, potato starch, microcrystalline cellulose, acacia, gelatin, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers.
  • a formulation suitable for oral administration can include lozenge forms, that can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier; as well as creams, emulsions, gels, and the like containing, in addition to the active ingredient, such carriers as are known in the art.
  • An aerosol formulation suitable for administration via inhalation also can be made.
  • the aerosol formulation can be placed into a pressurized acceptable propellant, such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • a formulation suitable for topical application can be in the form of creams, ointments, or lotions.
  • the dose administered to an animal, particularly a human, in the context of the present invention should be sufficient to effect a (protective) immune response in the infected individual over a reasonable time frame.
  • the dose will be determined by the potency of the particular vector or construct employed, the severity of the disease state, as well as the body weight and age of the infected individual.
  • the size of the dose also will be determined by the existence of any adverse side effects found to accompany the use of the particular vector or construct employed. It is always desirable, whenever possible, to keep adverse side effects to a minimum.
  • the dosage can be in unit dosage form.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of a vector or construct, alone or in combination with other antiviral agents, calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier, or vehicle.
  • the specifications for the unit dosage forms of the present invention depend on the particular compound or compounds employed and the effect to be achieved, as well as the pharmacodynamics associated with each compound in the host.
  • the dose administered should be an "effective amount” or an amount necessary to achieve an "effective level" in the individual patient.
  • the effective level is used as the preferred endpoint for dosing, the actual dose and schedule can vary, depending on individual differences in pharmacokinetics, drug distribution, and metabolism.
  • the "effective level” can be defined, for example, as the blood or tissue level desired in the patient that corresponds to a concentration of one or more vectors or constructs according to the invention, which produces the desired level of immune response or protective vaccinated state, in an assay predictive for clinical efficacy.
  • the "effective level” for use according to the present invention also can vary when the compositions of the present invention are used in combination with zidovudine or other known antiviral compounds or combinations thereof.
  • the skilled person can easily determine the appropriate dose, schedule, and method of administration for the exact formulation of the composition being used, in order to achieve the desired "effective level" in the individual patient.
  • One skilled in the art also can readily determine and use an appropriate indicator of the "effective level" of the agents of the present invention by a direct (e.g., analytical chemical analysis) or indirect (e.g., with surrogate indicators of viral infection, such as p24 or reverse transcriptase for treatment of AIDS or AIDS-like disease) analysis of appropriate patient samples (e.g., blood and/or tissues).
  • suitable animal models are available and have been widely implemented for evaluating the in vivo efficacy of various immunogens against HIV. Similar models for other viruses and infectious agents are also known to the skilled person. The models include mice, monkeys and cats.
  • mice models e.g., SCID, bg/nu/xid, NOD/SCID, SCID-hu, immunocompetent SCID-hu, bone marrow-ablated BALB/c
  • PBMCs peripheral blood mononuclear cells
  • lymph nodes fetal liver/thymus or other tissues
  • SIV simian immune deficiency virus
  • FV feline immune deficiency virus
  • suitable animal models are available and have been widely implemented for evaluating the in vivo efficacy of various immunogens against HIV. Similar models for other viruses and infectious agents are also known to the skilled person. These models can also be used to determine the safety of a vector for the purposes of validation of the vector system for clinical trials. An important application is the use of these animal models for biodistribution studies. Transduced cells, preferably but not limited to human cells, containing a vector are injected into a non-human animal model and the extent of distribution of the vector is determined by the presence of vector genetic material in animal tissue.
  • the absence of vector genetic material in animal tissue would mean that the vector does not autonomously replicate and spread to other cells and thus may be used in accord with the present invention.
  • the presence of vector in other cells would indicate that the vector was able to propagate beyond the original cells.
  • the vectors autonomously replicate they can be evaluated according to other criteria for safety, such as, but not limited to, the lack of replication in certain tissues or the level of replication in the animal.
  • the presence or absence of the vector could be determined by PCR, or by FACS analysis if the tested vector expresses a marker gene that can be visualized by FACS, but is not limited to such means of detection.
  • an amount of vector sufficient to achieve a tissue concentration of the administered vector or construct of from about 5 ⁇ g/kg to about 300 mg/kg of body weight is preferred, especially of from about 10 ⁇ g/kg to about 200 mg/kg of body weight.
  • the number of doses will vary depending on the means of delivery and the particular vector administered.
  • the treatment i.e., the administration of conditionally replicating or replication deficient constructs
  • the treatment is necessarily limited.
  • the pharmaceutical composition can be in the form of a medicament containing other pharmaceuticals, in conjunction with a vector or construct according to the invention, when used to therapeutically treat AIDS.
  • These other pharmaceuticals can be used in their traditional fashion (i.e., as agents to treat disease).
  • an antiretroviral agent be employed, such as, preferably, zidovudine.
  • additional pharmaceuticals include antiviral compounds, immunomodulators, immunostimulants, antibiotics, and other agents and treatment regimes (including those recognized as alternative medicine) that can be employed to treat AIDS.
  • Antiviral compounds include, but are not limited to, ddI, ddC, gancylclovir, fluorinated dideoxynucleotides, nonnucleoside analog compounds such as nevirapine ( Shih et al., PNAS, 88, 9878-9882 (1991 )), TIBO derivatives such as R82913 ( White et al., Antiviral Research, 16,257-266 (1991 )), BI-RJ-70 ( Shih et al., Am. J. Med., 90(Suppl. 4A), 8S-17S (1991 )) and the agents and regimens known to the skilled person as described above.
  • Immunomodulators and immunostimulants include, but are not limited to, various interleukins, CD4, cytokines, antibody preparations, blood transfusions, and cell transfusions.
  • Antibiotics include, but are not limited to, antifungal agents, antibacterial agents, and anti-Pneumocystis carinii agents.
  • virus-inhibiting compound with other anti-retroviral agents and particularly with known RT inhibitors, such as ddC, zidovudine, ddI, ddA, or other inhibitors that act against other HIV proteins, such as anti-TAT agents, will generally inhibit most or all replicative stages of the viral life cycle.
  • RT inhibitors such as ddC, zidovudine, ddI, ddA, or other inhibitors that act against other HIV proteins, such as anti-TAT agents.
  • ddC and zidovudine used in AIDS or ARC patients have been published.
  • a virustatic range of ddC is generally between 0.05 ⁇ M to 1.0 ⁇ M.
  • a range of about 0.005-0.25 mg/kg body weight is virustatic in most patients.
  • the dose ranges for oral administration are somewhat broader, for example 0.001 to 0.25 mg/kg given in one or more doses at intervals of 2, 4, 6, 8, and 12; etc., hr.
  • 0.01 mg/kg body weight ddC is given every 8 hr.
  • the other antiviral compound for example, can be given at the same time as a vector according to the invention, or the dosing can be staggered as desired.
  • the vector also can be combined in a composition. Doses of each can be less, when used in combination, than when either is used alone.
  • kits comprising components such as the vectors of the invention for use in the practice of the methods disclosed herein, where such kits may comprise containers, each with one or more of the various reagents (typically in concentrated form) utilized in the methods, including, for example, buffers and other reagents as necessary.
  • a label or indicator describing, or a set of instructions for use of, kit components in a method of the present invention, will also be typically included, where the instructions may be associated with a package insert and/or the packaging of the kit or the components thereof.
  • the vector-based strategies of the present invention are expected to be particularly beneficial, and more effective, against the elusive nature of certain pathogens, such as HIV.
  • the invention may be easily applied to other infectious diseases with equal effectiveness.
  • the advantage provided by the present invention is the production of a vaccine capable of reliably eliciting a robust cellular and humoral response to the antigens presented thereby, without requiring extensive research and development with each emerging disease.
  • the ability of conditionally replicating HIV based vectors to complement each other in culture and propagate for a limited period of time was investigated.
  • the first vector was an HIV-based vector containing all proteins necessary for replication, but lacking an envelope protein.
  • the second vector expressed the envelope protein from vesicular stomatitis virus (VSV-G). Plasmids were cotransfected into 12 x 10 6 cells 293F cells on a 1500 mm dish at 25 ⁇ g and 20 ⁇ g respectively. 24 hours post transfection, the supernatant was collected, aliquoted, and stored at -80 °C until use. Supernatant was diluted 5, 10, 50, 100, and 1000-fold in media, and added to 1 x 10 6 HeLa-tat cells in duplicate in 6 well plates.
  • Virus propagation was measured by p24 ELISA (ABI Labs, Kensington MD) on cell supernatants as a measure of virion production. A dose-dependent level of virion production was observed, and eventual diminution of complementation occurred ( Figure 5 ). Thus as expected the vectors replicated for a period of time before ending their replication cycles with cells that contain one of the two vector constructs. This is depicted in Figure 1 and provides an added advantage, or important safety feature, to the instant invention.
  • the termination of vector propagation indicates that regardless of the level of immune response that is generated, the vectors do not continue to replicate. Thus the vectors do not continue their spread after either generation of a protective immune response (immunity) or not.
  • the generation of a protective immune response can be tested by known assays, including but not limited to in vitro assays for cellular and antibodies as well as animal models of disease, such as simians for therapies targeting HIV.
  • vector propagation may be re-activated by the repeat use of the vector combination.
  • reactivation may be by the use of the individual vectors, optionally in virion form to superinfect cells that contain one or the other of the vectors originally used.
  • the first vector has all necessary components except a viral envelope protein which is provided by a second vector
  • the second vector capable of expressing the envelope protein may be packaged into viral particles and then introduced into cells of a subject in whom propagation of the two vectors has stopped. Infection of cells containing only the first vector by viral particles containing the second vector would reactivate propagation of the two vectors to further induce, or boost, the immune response.
  • this reactivation would again be limited in the extent of propagation in a manner analogous to that discussed and observed above.
  • the use of a different virion particle may be used.
  • a second vector used to reactivate or "boost" vector propagation may be modified such that the second vector is packaged into a viral particle displaying different antigens.
  • the same vector may be packaged into a particle displaying antigens D, E, and F to avoid the immune response in the subject.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Public Health (AREA)
  • Communicable Diseases (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biochemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • AIDS & HIV (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
EP08020274A 2003-09-09 2004-09-09 Approches basées sur un vecteur lentivirus pour la génération d'une réponse immunitaire au VIH chez les humains Withdrawn EP2055316A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US50166503P 2003-09-09 2003-09-09
EP04788661A EP1677832A1 (fr) 2003-09-09 2004-09-09 Approches a base de vecteurs de lentivirus permettant de generer une reponse immunitaire au virus de l'immunodeficience humaine

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
EP04788661A Division EP1677832A1 (fr) 2003-09-09 2004-09-09 Approches a base de vecteurs de lentivirus permettant de generer une reponse immunitaire au virus de l'immunodeficience humaine

Publications (1)

Publication Number Publication Date
EP2055316A1 true EP2055316A1 (fr) 2009-05-06

Family

ID=34273064

Family Applications (2)

Application Number Title Priority Date Filing Date
EP08020274A Withdrawn EP2055316A1 (fr) 2003-09-09 2004-09-09 Approches basées sur un vecteur lentivirus pour la génération d'une réponse immunitaire au VIH chez les humains
EP04788661A Withdrawn EP1677832A1 (fr) 2003-09-09 2004-09-09 Approches a base de vecteurs de lentivirus permettant de generer une reponse immunitaire au virus de l'immunodeficience humaine

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP04788661A Withdrawn EP1677832A1 (fr) 2003-09-09 2004-09-09 Approches a base de vecteurs de lentivirus permettant de generer une reponse immunitaire au virus de l'immunodeficience humaine

Country Status (6)

Country Link
US (3) US20050196381A1 (fr)
EP (2) EP2055316A1 (fr)
JP (1) JP2007505130A (fr)
AU (1) AU2004270275B2 (fr)
CA (1) CA2537953A1 (fr)
WO (1) WO2005023313A1 (fr)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007109131A2 (fr) * 2006-03-17 2007-09-27 Massachusetts Institute Of Technology Vecteurs lentiviraux qui fournissent une expression optimisee et une variegation reduite suivant la transgenese
US7847085B2 (en) * 2006-06-30 2010-12-07 New York University Recombinant HIV-1 gp120 immunogen with three different V3 loops from viruses of different clades
EP2222861B1 (fr) 2007-12-11 2017-12-06 The University of North Carolina At Chapel Hill Vecteurs rétroviraux modifiés dépourvus de sequence riche en polypurine (ppt)
US20120135034A1 (en) * 2009-03-13 2012-05-31 Boro Dropulic Non-Integrating Retroviral Vector Vaccines
EP2667893A2 (fr) * 2011-01-27 2013-12-04 Lentigen Corporation Vaccin de primovaccination et de vaccination de rappel avancé
EP3044339B1 (fr) 2013-09-10 2019-05-22 Mockv Solutions Procédés et kits permettant de quantifier l'élimination de fausses particules virales d'une solution purifiée
EP3031923A1 (fr) 2014-12-11 2016-06-15 Institut Pasteur Composition immunogène contre l'encéphalite japonaise à base de vecteurs lentiviraux
WO2023076180A2 (fr) 2021-10-25 2023-05-04 Genvivo, Inc. Compositions et méthodes d'administration thérapeutique ou vaccinale

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999004026A2 (fr) * 1997-07-18 1999-01-28 Chiron Corporation Vecteurs lentiviraux
US5866137A (en) 1990-10-12 1999-02-02 Benjamin Rovinski Self-assembled, non-infectious, non-replicating; immunogenic retrovirus-like particles comprising modified HIV genomes devoid of long terminal repeats and chimeric envelope glycoproteins
US6168953B1 (en) 1995-11-28 2001-01-02 The Johns Hopkins University School Of Medicine Genetic antiviral agents and methods for their use

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040063652A1 (en) * 1988-03-21 2004-04-01 Jolly Douglas J. Combination gene delivery vehicles
US5665577A (en) * 1989-02-06 1997-09-09 Dana-Farber Cancer Institute Vectors containing HIV packaging sequences, packaging defective HIV vectors, and uses thereof
GB8923123D0 (en) * 1989-10-13 1989-11-29 Connaught Lab A vaccine for human immunodeficiency virus
US5861282A (en) * 1989-10-16 1999-01-19 Whitehead Institute For Biomedical Research Non-infectious HIV particles and uses therefor
WO1992003537A1 (fr) * 1990-08-15 1992-03-05 Therion Biologics Corporation Particules de virus hybrides a replication defectueuse et auto-assemblees
US6297048B1 (en) * 1992-02-04 2001-10-02 Chiron Corporation Hepatitis therapeutics
US5834256A (en) * 1993-06-11 1998-11-10 Cell Genesys, Inc. Method for production of high titer virus and high efficiency retroviral mediated transduction of mammalian cells
US6013516A (en) * 1995-10-06 2000-01-11 The Salk Institute For Biological Studies Vector and method of use for nucleic acid delivery to non-dividing cells
US5741492A (en) * 1996-01-23 1998-04-21 St. Jude Children's Research Hospital Preparation and use of viral vectors for mixed envelope protein vaccines against human immunodeficiency viruses
FR2747046B1 (fr) * 1996-04-05 1998-06-19 Univ Paris Curie Nouveaux vaccins issus de plasmovirus
CA2267683A1 (fr) * 1996-10-04 1998-04-09 Demeter Biotechnologies, Ltd. Procede de traitement d'une contamination par le virus de l'immunodeficience
US6248721B1 (en) * 1997-04-09 2001-06-19 Lung-Ji Chang Method of using mouse model for evaluation of HIV vaccines
US6207455B1 (en) * 1997-05-01 2001-03-27 Lung-Ji Chang Lentiviral vectors
US5994136A (en) * 1997-12-12 1999-11-30 Cell Genesys, Inc. Method and means for producing high titer, safe, recombinant lentivirus vectors
US6121021A (en) * 1997-12-16 2000-09-19 Connaught Laboratories Limited Constitutive expression of non-infectious HIV-like particles
WO1999058726A1 (fr) * 1998-05-12 1999-11-18 Genecure Llc Vaccin de vih a deficience de replication
EP1059357A1 (fr) * 1999-06-09 2000-12-13 Universite Pierre Et Marie Curie Paris Vi Constructions rétrovirales capables de la réplication autonome ou semi-autonome, leur préparation et leurs utilisations pour le transfert de gènes
DE60140052D1 (de) * 2000-05-31 2009-11-12 Human Gene Therapy Res Inst Methode und zusammensetzungen für effizienten gentransfer durch transcomplementierende vektoren
US6627442B1 (en) * 2000-08-31 2003-09-30 Virxsys Corporation Methods for stable transduction of cells with hiv-derived viral vectors
JP2004524813A (ja) * 2000-09-22 2004-08-19 バイレクシス コーポレイション 改良された条件付けで複製するベクター類、それらの生成及び使用方法
US6712612B1 (en) * 2000-12-12 2004-03-30 Genecure Llc Safe and stable retroviral helper cell line and related compositions and methods
AU2002327338A1 (en) * 2001-07-25 2003-02-17 New York University Use of glycosylceramides as adjuvants for vaccines against infections and cancer

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5866137A (en) 1990-10-12 1999-02-02 Benjamin Rovinski Self-assembled, non-infectious, non-replicating; immunogenic retrovirus-like particles comprising modified HIV genomes devoid of long terminal repeats and chimeric envelope glycoproteins
US6168953B1 (en) 1995-11-28 2001-01-02 The Johns Hopkins University School Of Medicine Genetic antiviral agents and methods for their use
US6410257B1 (en) 1995-11-28 2002-06-25 The Johns Hopkins University School Of Medicine Methods to assay gene function with viral vectors
WO1999004026A2 (fr) * 1997-07-18 1999-01-28 Chiron Corporation Vecteurs lentiviraux

Non-Patent Citations (30)

* Cited by examiner, † Cited by third party
Title
"Centers for Disease Control and Prevention", HIV/AIDS SURVEILLANCE REPORT., vol. 13, 2001
"UNAIDS Joint United Nations Programme on HIV/AIDS", AIDS EPIDEMIC UPDATES, 2002
AMARA RR ET AL.: "Control of a mucosal challenge and prevention of AIDS by a multiprotein DNA/MVA vaccine", SCIENCE, vol. 292, 2001, pages 69 - 74
BEHRENS G.M.N.: "Stoll M. & Schmidt R.E. Lipodystrophy syndrome in HIV infection. What is it, what causes it, and how can it be managed?", DRUG SAF., vol. 23, 2000, pages 57 - 76
BRAUN ET AL: "Inhibition of Simian/Human Immunodeficiency Virus Replication in CD4<+> T Cells Derived from Lentiviral-Transduced CD34<+> Hematopoietic Cells", MOLECULAR THERAPY, ACADEMIC PRESS, SAN DIEGO, CA, US, vol. 12, no. 6, 1 December 2005 (2005-12-01), pages 1157 - 1167, XP005166238, ISSN: 1525-0016 *
BRENNER S ET AL: "Current developments in the design of onco-retrovirus and lentivirus vector systems for hematopoietic cell gene therapy", BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 1640, no. 1, 7 April 2003 (2003-04-07), pages 1 - 24, XP004417690, ISSN: 0167-4889 *
DEACON N.J. ET AL.: "Genomic structure of an attenuated quasi species of HIV-1 from a blood transfusion donor and recipients", SCIENCE, vol. 270, 1995, pages 988 - 991
DROPULIC B: "Lentivirus in the clinic.", MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY. DEC 2001, vol. 4, no. 6, December 2001 (2001-12-01), pages 511 - 512, XP008042363, ISSN: 1525-0016 *
DROPULIC BORO ET AL: "Establishing safety in the clinic for the first lentiviral vector to be tested in humans", BLOOD, vol. 104, no. 11, Part 1, November 2004 (2004-11-01), & 46TH ANNUAL MEETING OF THE AMERICAN-SOCIETY-OF-HEMATOLOGY; SAN DIEGO, CA, USA; DECEMBER 04 -07, 2004, pages 121A, XP008104344, ISSN: 0006-4971 *
GEA-BANACLOCHE, J. C. ET AL.: "Maintenance of large numbers of virus-specific CDB+ T cells in HIV-infected progressors and long-term nonprogressors", J. IMMUNOL., vol. 165, 2000, pages 1082 - 1092
LEARMONT J.C. ET AL.: "Immunologic and virologic status after 14 to 18 years of infection with an attenuated strain of HlV-1. A report from the Sydney Blood Bank Cohort", N. ENGL. J. MED, vol. 340, 1999, pages 1715 - 1722
LEVINE BRUCE L ET AL: "Gene transfer in humans using a conditionally replicating lentiviral vector", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 103, no. 46, November 2006 (2006-11-01), pages 17372 - 17377, ISSN: 0027-8424 *
LEVINE BRUCE L ET AL: "Gene transfer in humans using a conditionally replicating lentiviral vector.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 14 NOV 2006 LNKD- PUBMED:17090675, vol. 103, no. 46, 14 November 2006 (2006-11-14), pages 17372 - 17377, XP008153054, ISSN: 0027-8424, DOI: 10.1073/pnas.0608138103 *
LITTLE S.J. ET AL.: "Antiretroviral-drug resistance among patients recently infected with HIV", NEW ENG. J. MED, vol. 347, 2002, pages 385 - 394
LUCAS G.M.; CHAISSON R.E.; MOORE R.D.: "Highly active antiretroviral therapy in a large urban clinic: risk factors for virologic failure and adverse drug reactions", ANN. INT. MED, vol. 131, 1999, pages 81 - 87
MAX B.; SHERER R..: "Management of the adverse effects of antiretroviral therapy and medication adherence", CLIN. INFECT. DIS., vol. 30S, 2000, pages 96 - 116
MITTA B ET AL: "Advanced modular self-inactivating lentiviral expression vectors for multigene interventions in mammalian cells and in vivo transduction", NUCLEIC ACIDS RESEARCH, OXFORD UNIVERSITY PRESS, SURREY, GB, vol. 30, no. 21, 1 November 2002 (2002-11-01), pages e113, XP002321925, ISSN: 0305-1048 *
NIJUIS M.; DEEKS S.; BOUCHER C.: "Implications of antiretroviral resistance on viral fitness", CURR. OPIN. INFEC. DIS., vol. 14, 2001, pages 23 - 28
POWDERLY WG.: "Long-term exposure to lifelong therapies", J. ACQ. IMM. DEF SYND, vol. 29S, 2002, pages 28 - 40
SCHLATTER S ET AL: "SAMY, a novel mammalian reporter gene derived from Bacillus stearothermophilus alpha-amylase", GENE, ELSEVIER, AMSTERDAM, NL, vol. 282, no. 1-2, 9 January 2002 (2002-01-09), pages 19 - 31, XP004334702, ISSN: 0378-1119 *
SCIENTIFIC JOURNAL NATURE, January 2002 (2002-01-01)
SHIH ET AL., AM. J. MED., vol. 90, no. 4A, 1991, pages 8S - 17S
SHIH ET AL., PNAS, vol. 88, 1991, pages 9878 - 9882
SIRIN O ET AL: "Regulating gene expression using self-inactivating lentiviral vectors containing the mifepristone-inducible system", GENE, ELSEVIER, AMSTERDAM, NL, vol. 323, 24 December 2003 (2003-12-24), pages 67 - 77, XP004477031, ISSN: 0378-1119 *
STATHOPULOS P B: "Taking the good out of the bad: lentiviral-based gene therapy of the hemoglobinopathies", BIOTECHNOLOGY ADVANCES, ELSEVIER PUBLISHING, BARKING, GB, vol. 21, no. 6, 1 September 2003 (2003-09-01), pages 513 - 526, XP004448474, ISSN: 0734-9750 *
TODD S ET AL: "HIV protease as a target for retrovirus vector-mediated gene therapy", BIOCHIMICA ET BIOPHYSICA ACTA - PROTEIN STRUCTURE AND MOLECULAR ENZYMOLOGIE, ELSEVIER SCIENCE BV, AMSTERDAM, NL, vol. 1477, no. 1-2, 7 March 2000 (2000-03-07), pages 168 - 188, XP004278893, ISSN: 0167-4838 *
TURNER B.J.: "Adherence to antiretroviral therapy by human immunodeficiency virus-infected patients", J. INFEC. DIS., vol. 185S, 1993, pages 145 - 151
VIGOUROUX, C. ET AL.: "Adverse metabolic disorders during highly active antiretroviral treatments (HAART) ofHIV disease", DIAB. METAB., vol. 25, 1999, pages 385 - 392
WHITE ET AL., ANTIVIRAL RESEARCH, vol. 16, 1991, pages 257 - 266
WYAND S. ET AL.: "Protection by live, attenuated simian immunodeficiency virus against heterologous challenge", J VIROL., vol. 73, 1999, pages 8356 - 8363

Also Published As

Publication number Publication date
WO2005023313A1 (fr) 2005-03-17
CA2537953A1 (fr) 2005-03-17
US20050196381A1 (en) 2005-09-08
JP2007505130A (ja) 2007-03-08
AU2004270275A1 (en) 2005-03-17
EP1677832A1 (fr) 2006-07-12
US20060281128A1 (en) 2006-12-14
AU2004270275B2 (en) 2010-02-18
US20150030627A1 (en) 2015-01-29

Similar Documents

Publication Publication Date Title
US20150030627A1 (en) Trans-complementing, replication deficient lentiviral vectors and methods for making and using them
JP6480028B2 (ja) レンチウイルス遺伝子導入ベクター及びそれらの医薬品への適用
Schell et al. Significant protection against high-dose simian immunodeficiency virus challenge conferred by a new prime-boost vaccine regimen
KR20090034297A (ko) 인플루엔자에 대한 항바이러스제 및 백신
AU2009228183A1 (en) Lentivirus-based immunogenic vectors
RU2301260C2 (ru) Вирусные векторы с зависимой от условий репликацией и их применение
US20040033595A1 (en) Conditionally replicating vectors for inhibiting viral infections
DK2020444T3 (en) DEFECTIVE NON-INTEGRATIVE LENTIVIRAL TRANSMISSION VECTORS FOR VACCINES
JP2007505130A5 (fr)
US20050208072A1 (en) Preventive and therapeutic aids vaccines
Lemiale et al. Lentiviral vectors for HIV disease prevention and treatment
Anraku et al. Kunjin replicon-based simian immunodeficiency virus gag vaccines
EP1255849A2 (fr) Rhabdovirus recombines utiles en tant que vaccins a virus vivants diriges contre des virus a deficit immunitaire
US9566329B2 (en) Live, attenuated rubella vector to express vaccine antigens
Jia et al. Single-cycle SIV: a novel AIDS vaccine approach
AU2001293075B2 (en) Conditionally replicating viral vectors and their use
McKenna Characterization and immunogenicity of recombinant rhabdoviral vaccine vectors expressing HIV-1 and SIV envelope proteins
Malm Assessing the Immunogenicity of GTU®-based HIV-1 Multigene DNA Vaccines in Murine Models
WO2002098457A2 (fr) Rhabdovirus recombinants en tant que vaccins antiviraux actifs pour des virus de l&#39;immunodeficience
AU2001293075A1 (en) Conditionally replicating viral vectors and their use
AU2002258639A1 (en) Improved conditionally replicating vectors for inhibiting viral infections

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AC Divisional application: reference to earlier application

Ref document number: 1677832

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PL PT RO SE SI SK TR

17P Request for examination filed

Effective date: 20091015

17Q First examination report despatched

Effective date: 20091112

AKX Designation fees paid

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PL PT RO SE SI SK TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20120611